You are on page 1of 11

Current Nutrition Reports (2021) 10:47–57

https://doi.org/10.1007/s13668-020-00347-9

MATERNAL AND CHILDHOOD NUTRITION (AC WOOD, SECTION EDITOR)

Artificial Pancreas Technology Offers Hope for Childhood Diabetes


Melissa J. Schoelwer 1 & Mark D. DeBoer 1

Accepted: 20 December 2020 / Published online: 7 January 2021


# The Author(s), under exclusive licence to Springer Science+Business Media, LLC part of Springer Nature 2021

Abstract
Purpose of Review Here, we provide a review of recent advancements in closed-loop or automated insulin delivery systems
commonly referred to as the “artificial pancreas” (AP) for the management of type 1 diabetes (T1D).
Recent Findings The use of hybrid closed-loop devices in children and adolescents with T1D consistently increases time in the
target glucose range (70–180 mg/dL) without increasing the risk of hypoglycemia. Although hybrid closed-loop systems are able
to maintain fairly tight glycemic control overnight, daytime control remains challenging, primarily due to meals and exercise, and
careful carbohydrate counting and meal boluses remain essential components of optimal diabetes control. Bihormonal and fully
automated AP systems remain investigational at this time.
Summary While commercially available hybrid closed-loop AP systems improve glycemic control in children with T1D, more
work is needed to achieve a fully automated AP system and decrease the burden of using diabetes technology.

Keywords Artificial pancreas . Type 1 diabetes . Closed-loop control . Automated insulin delivery . Pediatric

Introduction limiting exposure to hypoglycemia. The benefits of intensive


management for the prevention of long-term complications
Type 1 diabetes (T1D) is an autoimmune disease that results were first demonstrated in the landmark Diabetes Control
in destruction of the pancreatic islet cells and the inability to and Complications Trial (DCCT) published in 1993 [2].
produce insulin, an essential hormone for glucose metabolism. Glycemic targets for children with T1D have become more
T1D often presents in childhood and the incidence appears to rigorous over time, and the HbA1c goal is now < 7%
be rising worldwide [1]. The management of T1D is compli- (53 mmol/mol) for all children regardless of age [3, 4].
cated and is centered on frequent monitoring of blood glucose, While the majority of children with T1D are unable to meet
careful carbohydrate counting, and subcutaneous insulin ad- the HbA1c target with current therapies [5•], advancements in
ministration. Insulin can be administered either with multiple the field of diabetes technology have shown significant prom-
daily subcutaneous injections with a syringe or a pen device or ise in improving glycemic control and decreasing the burden
via continuous infusion with an insulin pump, a device worn of diabetes management. The remainder of this review will
outside the body that infuses rapid-acting insulin through a focus on closed-loop control or automated insulin delivery
subcutaneous cannula that is inserted every 2–3 days. technology, otherwise commonly referred to as “artificial pan-
The primary goal of diabetes management is to prevent the creas” (AP) systems.
development of macro- and microvascular complications such
as cardiovascular disease, retinopathy, nephropathy, and neu-
ropathy by achieving near-normal glycaemia while also Search Strategy and Selection Criteria

This article is part of the Topical Collection on Maternal and Childhood A PubMed search was done using the terms “artificial pancre-
Nutrition as,” “closed-loop control,” or “automated insulin delivery” in
combination with “type 1 diabetes.” The search was limited to
* Mark D. DeBoer the past 5 years (2015–present). Articles were chosen for in-
deboer@virginia.edu
clusion based on study design and applicability to this review
1
Division of Pediatric Endocrinology and Center for Diabetes
with preference given to pediatric studies and pivotal trials
Technology, University of Virginia, 1215 Lee Street, resulting in FDA-approved AP systems. Relevant literature
Charlottesville, VA 22903, USA was also obtained from the reference lists of chosen articles.
48 Curr Nutr Rep (2021) 10:47–57

Artificial Pancreas Basics CGM-based glycemic outcomes are now widely reported
in the AP literature to detect improvements in glycemic con-
There are three key components of an AP system: an insulin trol outside of the customary HbA1c. Clinical targets for
pump, a continuous glucose monitor (CGM) or “sensor,” and CGM-based outcomes were recently published, with the goal
a control algorithm. The majority of AP systems use insulin time in range (TIR, 70–180 mg/dL) set to ≥ 70% and time
only; however, dual-hormone devices are also under investi- spent in hypoglycemia (< 70 mg/dL) to ≤ 4% [10].
gation, primarily with the addition of glucagon. The algorithm
uses information from the CGM (current glucose and glucose
rate of change) along with the calculated insulin on board
Control Algorithms
(active insulin) to automatically adjust insulin delivery to keep
the blood glucose within a desired target range [6]. AP sys-
The control algorithms used in AP systems take into ac-
tems that are presently commercially available are considered
count various inputs including CGM data, insulin param-
hybrid closed-loop (HCL) systems as they still require that the
eters programmed into the pump, total daily insulin, and
user count and enter the grams of carbohydrates consumed
insulin on board and use predictive models to guide insu-
with each meal or snack for optimal performance.
lin dosing decisions. Three different control strategies
Additional input about exercise and sleep might also be en-
have been employed in AP systems: proportional, inte-
tered depending on the system. In contrast, a fully closed-loop
gral, derivative (PID) control, model predictive control
system would not require any input from the user about meals
(MPC), and fuzzy logic (FL) [6, 11]. A PID algorithm
or exercise.
adjusts basal insulin delivery in response to real-time glu-
cose readings to the target glucose whereas a MPC algo-
rithm adjusts insulin delivery based on its prediction of a
future blood glucose value. A controller using FL also
Continuous Glucose Monitors
uses CGM data but tries to imitate clinical decisions when
altering insulin dosing. Each control algorithm is unique
The development of accurate CGM devices over the past
and most controllers use a combination of these tech-
20 years was crucial for the advancement of AP technology.
niques. Early control algorithm platforms were indepen-
CGM are inserted under the skin and are typically worn for 7–
dently located on a computer or smartphone, wirelessly
14 days depending on the device. They measure glucose in the
receiving data from the CGM and communicating with
interstitial fluid rather than in the blood, which results in a 5–
the insulin pump. Over time, these controllers have been
10-min delay when comparing with a blood glucose reading.
integrated into insulin pumps, allowing for greater sim-
The accuracy of commercial sensors is measured by the mean
plicity and improved connectivity.
absolute relative difference (MARD), which compares a sen-
sor reading with a blood glucose measurement [7]. Accuracy
has improved significantly over time. Early sensors required
multiple daily calibrations with a blood glucose meter value to Artificial Pancreas Development
increase precision. However, many current sensors have a
MARD < 10%, a threshold that is largely considered accurate First-Generation Systems
enough to make insulin dosing decisions, with few to no cal-
ibrations needed [8]. The first commercially available AP systems focused solely
The data from some CGM can be wirelessly transmitted to on limiting hypoglycemia, which is a significant concern for
another device such as a smartphone or insulin pump for 24-h patients with T1D, particularly overnight [12]. The Medtronic
real-time monitoring of glucose trends, a feature particularly 530G Threshold Suspend pump released in 2013 stopped in-
appreciated by parents of children with T1D. CGM use has sulin delivery during times of hypoglycemia, referred to as
increased dramatically in the pediatric age range over the last low glucose suspend (LGS) system [13, 14]. This was quickly
few years, especially in children under the age of 12 years [5•]. followed by the ability to stop or decrease insulin delivery
The use of CGM is associated with improved glycemic out- prior to the development of frank hypoglycemia, known as
comes, regardless of insulin delivery modality used for diabe- predictive low glucose suspend (PLGS). The Medtronic
tes management, along with a reduction in severe hypoglyce- 640G became available in 2015 [15, 16] followed by
mia and diabetic ketoacidosis [5, 9]. At the time of this pub- Tandem Basal-IQ in 2018 [17], both of which utilize PLGS
lication, there are four CGM devices commercially available algorithms. These devices were found to be effective in
in the USA: the Medtronic Guardian 3, the Dexcom G6, the preventing hypoglycemia without increasing hyperglycemia
long-term implantable Senseonics Eversense, and a flash and marked a significant milestone in automated insulin de-
CGM known as the Freestyle Libre. livery [17, 18].
Curr Nutr Rep (2021) 10:47–57 49

FDA-Approved Hybrid Closed-Loop Systems over the course of 3 months [21••]. The mean TIR achieved in
this trial was 65%, which was a significant increase from
Two hybrid closed-loop systems are commercially available baseline. Additionally, less time in hypoglycemia and a lower
in the USA at the time of this report, the Medtronic 670G and HbA1c was reported with use of the system. Auto mode was
the Tandem Control-IQ. Key similarities and differences of in use 81% of the time during this study.
these systems are outlined in Table 1. Since FDA approval, a number of studies have been con-
ducted on real-world outcomes using the Medtronic 670G
system. Stone et al. reported an increase in TIR in users in
auto mode compared to manual mode (73.3 vs. 66.0%) [22].
Medtronic 670G Despite encouraging reports of improved glycemic outcome,
discontinuation of the system appears to be fairly common,
The Medtronic MiniMed 670G system, consisting of the particularly in adolescents. One study in children noted a 37%
MiniMed 670G insulin pump, the Guardian 3 CGM, and a discontinuation rate over the course of 6 months along with a
“SmartGuard” algorithm, was the first on the market in 2018 decrease in time spent in auto mode and sensor wear over time
and is now approved by the FDA for use in patients down to [23]. It is likely that the burden of wearing multiple devices
7 years of age [19]. When in auto mode, the system adjusts and keeping the technology connected contributes to AP
basal insulin delivery every 5 min, targeting blood glucose of discontinuation.
120 mg/dL. While it requires programming with standard Only one publication of the use of the Medtronic 670G
pump parameters such as carbohydrate ratios and correction system was found in children under the age of 7 years, a
factors, it uses total daily insulin over the preceding 2–6 days retrospective review of 16 children (average age 4.3 years)
to determine these parameters while in auto mode. The at one center using the device off-label for 3–12 months
Guardian sensor requires at least 2 calibrations each day, al- [24]. The authors reported a significant increase in TIR when
though 3–4 calibrations per day are recommended for greatest using auto mode from 42.8 to 56.2% along with a reduction in
accuracy, and can be worn for up to 7 days. HbA1c (7.9 vs. 7.4%); however, significantly more time was
A number of feasibility, safety, and efficacy trials led up to spent in hypoglycemia (1.3 vs. 2.4%).
the phase 3 outpatient pivotal trials resulting in FDA approval
of this device. The first multicenter pivotal trial published by
Garg et al. in 2017 demonstrated increased TIR in adolescents
and adults using the system in auto mode for 3 months com- Tandem Control-IQ
pared to 2 weeks of use in manual mode [20]. Notably in that
trial, adolescents experienced more difficulty staying in auto The second FDA-approved hybrid closed-loop system is
mode than adults (75.8 vs. 88%, respectively). known as Tandem Control-IQ, which combines a Tandem
Forlenza and colleagues published the second multicenter t:slim X2 pump with the Dexcom G6 CGM. Unique features
pivotal trial in 2019 in children 7–13 years of age, also done of this system include its ability to give automated correction

Table 1 Comparison of FDA-approved artificial pancreas systems

Medtronic 670G Tandem Control-IQ Medtronic 770G

Ages approved 7 years and older 6 years and older 2 years and older
Control algorithm PID MPC PID
CGM used Guardian 3 Dexcom G6 Guardian 3
Basal insulin delivery Calculated every 5 min, irrespective Modified based on programmed basal rate Calculated every 5 min, irrespective
of programmed rate of programmed rate
Automated corrections No Yes No
Manual corrections Calculated by algorithm Uses programmed correction factor Calculated by algorithm
Target range 120 mg/dL 112.5–160 mg/dL depending on mode 120 mg/dL
Active insulin time Can be programmed Automatically set to 5 h, cannot be changed Can be programmed
Exercise mode feature No Yes No
Sleep mode feature No Yes No
Remote monitoring No Yes Yes
capability
50 Curr Nutr Rep (2021) 10:47–57

boluses in addition to modifying basal insulin delivery as well control group (Fig. 1). Importantly, time in closed-loop in
as intensifying glucose control overnight through a lower tar- both of these studies was high (> 90%).
get when in “sleep mode.” Real-world data of use of the Control-IQ system in 1435
Large randomized clinical trials of the Control-IQ system participants aged 14 years and older was recently published
were recently published in children, adolescents, and adults, and documented improvements both in glycemic and psycho-
demonstrating an increase in TIR compared to standard social outcomes [27]. In addition to high TIR on Control-IQ
therapy (sensor-augmented pump) in all age groups studied over the 1-month study (median TIR 78.2% and 79.2% at each
[25, 26]. Adolescents and adults with T1D increased TIR study point), the participants reported high satisfaction and
from 61 ± 17% at baseline to 71 ± 12% over the course of trust in the system.
6 months. In addition, mean HbA1c was significantly lower
(− 0.33%) in the closed-loop group compared to the control
group using sensor-augmented pumps after 6 months of use. Medtronic 770G
Similar findings were reported in children 6–12 years of age
who achieved a mean TIR of 67 ± 10% over 16 weeks of The FDA recently approved the Medtronic 770G system in
treatment with Control-IQ compared to 55 ± 13% in the September 2020 and it is expected to be commercially

Fig. 1 Percent time-in-target-


range (TIR) during a randomized
controlled trial (RCT) of an
artificial pancreas (AP) system vs.
usual care among school-aged
children age 6–12 years. Data
shown are from an RCT of the
Tandem Control-IQ system
demonstrating increased TIR 70–
180 mg/dL throughout the study
(a). This increase in TIR was
driven by improvements
overnight, a strength of AP
systems compared to usual care.
From N Engl J Med 2020;
383:836–845, used by permission
Curr Nutr Rep (2021) 10:47–57 51

available by the time of this publication. Updated features control groups are lacking [31]. In addition, the majority of
include the ability to monitor CGM data remotely using a articles reporting about DIY systems combine multiple types
smartphone app and the ability to do software updates to the of DIY systems, though each system likely has different levels
algorithm directly onto the pump, features that are also present of safety, efficacy, and acceptability. The lack of regulation of
in the Control-IQ system. CGM calibrations continue to be these devices is particularly cogent to their use in children who
recommended 2–4 times daily for this system. While it is in most cases do not have a complete understanding of the
approved down to 2 years of age, there is a minimum total potential risks. It is also possible that there may be a lower
daily insulin requirement of 8 units. degree of willingness among families using these devices to
The follow-up to the 770G, known as the 780G, is already report negative outcomes or adverse events given the stakes of
under development as well. There are a few reports in the providing untested algorithms for use in their children. With
literature of “enhanced HCL” Medtronic systems that have recent and on-going approval of commercially available AP
tested out several iterations of the upgraded algorithm [28]. systems, it remains unclear whether DIY systems will contin-
A small, 1-month feasibility trial in 12 adults showed that TIR ue in use or whether the majority of users will opt for FDA-
increased (85.3 vs. 75% at baseline) but time spent in hypo- approved systems.
glycemia also increased (4.4 vs. 3%) [28]. Time spent in
closed loop was high with few auto mode exits, a significant
improvement over the 670G experience. A small camp study Artificial Pancreas Systems
in adolescents reported similar TIR between users of the 670G Under Development
and an enhanced HCL system, although there were fewer auto
mode exits and alarms in the experimental group [29]. Similar There are multiple other insulin-only HCL AP systems that
findings were reported in a small adult study using an en- are under development and are being studied in clinical trials.
hanced system [30]. Insulet has been testing an AP algorithm for use in the
Omnipod patch insulin pump, which is a popular pump brand
in young children, as it does not have tubing. Pivotal trials for
“Do-It-Yourself” Artificial Pancreas Systems this system are underway, but it is not FDA approved at the
time of this report. Lily, Inc., has also published data from a
Over the past 5–10 years, multiple “Do-It-Yourself” (DIY) trial of an AP system developed by their company. A summa-
artificial pancreas systems have become available through ry of selected AP studies over the past 3 years, both those that
open-source platforms [31]. These systems utilize existing are commercially available and under development, is pre-
CGM and insulin pump products and connect them using sented in Table 2.
computer algorithms on a smartphone to create a closed-
loop system. The design of these systems has been driven in Dual-Hormone Artificial Pancreas Systems
large part by impatience among individuals in the T1D com-
munity with the long time required for regulatory approval for While the majority of AP systems have focused solely on
commercially available AP systems, as well as a desire to modulating insulin delivery to achieve improvements in
individualize therapy for a user’s needs [32]. Unlike tradition- TIR, a smaller number of systems have been developed that
al research-based approaches that require oversight by the infuse other hormones in addition to insulin. The most preva-
FDA and other regulatory agencies, these DIY systems have lent of these is glucagon, which is used to raise glucose levels
not had the same degree of rigorous testing and do not require acutely to avoid hypoglycemia. Damiano et al. from Boston
reporting of adverse events, leaving the safety of these sys- University developed such a system that has been reported in
tems less clear. multiple pediatric trials [35, 36]. These systems utilized a
In observational studies, parents who have chosen to use Dexcom CGM sensor that sends signals to a smartphone run-
these systems for insulin management in their children express ning the AP controller, which then sent infusion commands to
a high degree of satisfaction, and pre-/post-assessments of two separate insulin pumps—one delivering insulin and one
glycemic control on these systems compared to prior manage- glucagon. The studies reported thus far utilized a preparation
ment for individual users have reported a higher degree of TIR of glucagon that had to be replaced daily.
and less hypoglycemia [31, 33, 34]. One study reported an One of these dual-hormone studies evaluated adolescents
improvement in HbA1c among users from 6.91 to 6.27% 12–20 years old at a diabetes summer camp and was com-
(p < 0.001) and in TIR from 64.2 to 80.7% [33]—demonstrat- pared to usual care, each used for 5 days in a randomized
ing that in many cases, families using DIY systems were al- crossover study design [35]. On the dual-hormone system,
ready well controlled before use. TIR increased to 75.9% ± 7.9 from 64.5% ± 14 (p < 0.001)
It should be noted that most available data on DIY efficacy on usual care without significant change in time < 70 (6.1%
are self-reported and that randomized trials of these systems or and 7.6%). During dual-hormone AP use, adolescents
52 Curr Nutr Rep (2021) 10:47–57

Table 2 Review of select AP literature published within the past 3 years

Study AP system Study design Population Study length Results

Single hormone
Ekhlaspour et al. Tandem RCT N = 48 2-day ski camp Higher TIR in HCL group (66.4 vs. 53.9%) during
(2019) [52] Control-IQ Age 6–18 years intensive winter sport activities. Rates of
hypoglycemia were similar between both
groups.
Forlenza et al. Tandem RCT N = 24 3 days TIR was higher in the HCL group (71 vs. 52.8%)
(2019) [53] Control-IQ Age 6–12 years with similar rates of hypoglycemia.
Brown et al. Tandem RCT N=168 6 months TIR was higher in the HCL group compared to
(2019) [25••] Control-IQ Age 14–71 years control group (71 vs. 61%). Time in closed loop
was 90%.
Schoelwer et al. Tandem RCT N = 18 1 week Initializing HCL with simple “MyTDI”
(2020) [54] Control-IQ Age 12–18 years parameters based on total daily insulin resulted
in similar TIR as using home insulin pump
parameters in adolescents
Breton et al. Tandem RCT N = 101 4 months Mean TIR was higher in HCL than the control
(2020) [26••] Control-IQ Age 6–13 years group (67 vs. 55%). Median percentage of time
in closed loop was 93%.
Pinsker et al. Tandem Observational N = 1435 1 month Real-world use of Control-IQ showed most
(2020) [27] Control-IQ Mean age participants met CGM goal of > 70% TIR with
45.5 years only 1.7% hypoglycemia. Median TIR was
78.2% at T1 and 79.2% at T2.
Kovatchev et al. Tandem Randomized N = 80 8 months Use of HCL only in the evenings and overnight
(2020) [55] Control-IQ crossover Age 18–69 years reduced hypoglycemia compared to SAP and
achieved most of the glycemic benefits of 24-7
HCL
Forlenza et al. Medtronic 670G Observational N = 105 3 months At the end of the study, HbA1c decreased from 7.9
(2019) [21••] Age 7–13 years to 7.5% and TIR increased from 56.2 to 65%.
Median use of auto mode was 81%.
Lal et al. (2019) Medtronic 670G Observational N = 79 12 months Auto mode use was associated with lower HbA1c;
[56] Age 9–61 years however, 33% of 670G users discontinued use
by 12 months.
Messer et al. Medtronic 670G Observational N = 92 6 months 30% discontinued use of HCL by the end of the
(2020) [57] Age 8–25 years study. Higher baseline HbA1c predicted
discontinuation.
Berget et al. Medtronic 670G Observational N = 92 6 months TIR increased from 50.7% at baseline to 56.9%
(2020) [23] Mean age and HbA1c decreased from 8.7 to 8.4% at
15.7 ± 3.6 years 6 months; however, use of auto mode
decreased over time (only 51.2% at 6 months).
de Bock (2018) Medtronic RCT N = 12 1 week camp TIR was not significantly different between the
DTT [29] Enhanced HCL Age 13–17 years control group (standard 670G) and the e-HCL
users (75.85 vs.74.32%). There were 2.1
alarms/day in the control group compared to
0.26 in experimental group. Auto mode exits
were less frequent in the experimental group.
Lee et al. (2019) Medtronic Observational N = 12 1 week supervised TIR was higher with use of the e-HCL (85.3 vs.
[28] Enhanced HCL Median age hotel followed 75%). Time < 70 mg/dL also increased (4.4 vs.
48 years by 3 weeks of 3%). Time in closed loop was high (99.98%).
home use
Paldus et al. Medtronic Randomized N = 11 2 weeks TIR and mean glucose favored e-HCL but did not
(2019) [30] Enhanced HCL crossover reach statistical significance. e-HCL use
significantly decreased CL exits and alerts.
Tauschmann Cambridge RCT N = 86 3 months TIR was higher in the HCL group compared to the
et al. (2018) FlorenceM Age 6 years and control group (65% vs. 54%) and HbA1c was
[42] older (median lower (7.4% vs. 7.7%)
age 21 years)
Tauschmann Cambridge Randomized N = 24 6 weeks No difference in TIR when using closed loop with
et al. (2019) FlorenceM crossover Age 1–7 years standard insulin vs. diluted insulin (70 vs. 72%)
[58]
Curr Nutr Rep (2021) 10:47–57 53

Table 2 (continued)

Study AP system Study design Population Study length Results

Buckingham Omnipod Horizon Observational N = 12 54 h, supervised Safety of algorithm documented during


et al. (2018) Mean age hotel overestimated, missed, and extended meal
[59] 35.4 years boluses.
Buckingham Omnipod Horizon Observational N = 58 36 h, inpatient Primary safety endpoints of hypoglycemia
et al. (2018) Age 6–65 years < 70 mg/dL: 2% in children and adolescents.
[60] TIR during HCL was 72.6% for adolescents
and 70.1% for children.
Forlenza et al. Omnipod Horizon Observational N = 12 54-h supervised Subjects completed two > 30 min moderate
(2019) [61] Mean age hotel intensity exercise activities. Mean TIR was
36.5 years 85.1% with 1.4% time spent < 70 mg/dL (no
hypoglycemia overnight).
Sherr et al. (2020) Omnipod Horizon Observational N = 36 5 days, supervised TIR increased significantly for adolescents (79 vs.
[62] Age 8–48 years hotel 60.6%) and children (69.2 vs. 54.9%) compared
to standard therapy
Ekhlaspour et al. Bionic Pancreas Observational N = 13 3 weeks TIR 69% for static set-point and 72% for dynamic
(2019) [63] Age 19–37 years set-point (not significantly different)
Christiansen Lilly HCL Observational N = 20 3 days TIR on the system was 81.2% overall, 85.2%
(2020) [64] Mean age outside of challenges (meal and exercise), and
44.7 years 97.3% overnight.
Dual hormone
Castle et al. Insulin + Randomized N = 20 4 days Addition of glucagon with automated exercise
(2018) [65] glucagon crossover Age 21–45 years detection reduced hypoglycemia (3.4 vs. 8.3%
during exercise; 1.3 vs. 2.8% for entire study)
Haidar et al. Insulin + Randomized N = 28 Three 24-h Rapid insulin +pramlintide system increased TIR
(2020) [39] pramlintide crossover Mean age 25 years inpatient visits compared to insulin only (84 vs. 74%) due to
improved daytime control. Most common AE
from pramlintide was gastrointestinal issues.

AP, artificial pancreas; RCT, randomized controlled trial; TIR, time in rage (70–180 mg/dL); HCL, hybrid closed loop; CGM, continuous glucose
monitor; SAP, sensor-augmented pump; e-HCL, enhanced hybrid closed-loop; AE, adverse event

received a mean of 0.72 mg of glucagon (i.e., the majority of a amylin, which is secreted by pancreatic beta cells post-
standard rescue dose of 1 mg) each day. A similar study in prandially, resulting in slower gastric emptying and inhibition
children 6–11 years old found that TIR in this group on the of glucagon. Studies in adults have demonstrated that
dual-hormone system (vs. usual care) increased to 80.6% ± pramlintide-insulin systems produce improvements over insu-
7.4 from 57.6% ± 14.0, with a decrease in time < 70 mg/dL lin alone (TIR 84 vs. 74%), but these have not been tested in
(2.9 vs. 6.1%) [36]. These younger children received a mean pediatrics [39].
of 0.36 mg of glucagon daily. More recently, this research
group also published a pilot study that directly compared an Mobile Interoperable Closed-Loop Platforms
insulin-only with the dual-hormone system among adults with
cystic-fibrosis-related diabetes, finding similar glycemic con- The currently available AP systems run from algorithms em-
trol (TIR 70–180 mg/dL for glucagon-insulin system vs. in- bedded into the insulin pump; however, a potential alternative
sulin only; 80% ± 10 vs. 76% ± 9, p not significant) [37]. would be to run the system on a mobile platform using a
Another study in adults found that while the glucagon- smartphone. As previously discussed, early AP research was
insulin provided higher TIR during use compared to usual conducted in this manner and the first platform developed, the
care (78.4 vs. 61.9%), a visual analogue scale revealed signif- Diabetes Assistant (DiAS), was developed at the University of
icantly higher scores for nausea during dual-hormone use Virginia [40, 41]. Benefits of using a smartphone include that
[38•]. Overall, given that dual-hormone systems (as compared they are widely available, easy to upgrade, can offer a more
to insulin-only AP systems) appear to provide similar glyce- detailed user interface, and could potentially work with mul-
mic control but more nausea, it remains unclear what their role tiple different CGM and insulin pump devices for user
will be in pediatric AP control in the future. customizability. However, inconsistent wireless connectivity
Other researchers assessing dual-hormone AP systems may be a key limitation. Multiple mobile closed-loop plat-
have co-infused insulin and pramlintide, an analogue of forms are currently under investigation, FlorenceM
54 Curr Nutr Rep (2021) 10:47–57

(Cambridge algorithm), inControl AP, and Interoperable The development of a fully automated AP system remains
Artificial Pancreas System (iAPS). These systems have been the goal given the burden of carbohydrate counting and sig-
studied in clinical trials of varying sizes, and have also shown nificant contribution of missed meal boluses to poor glycemic
promise in improving glycemic control [42–44]. control. A few small studies evaluating fully automated insu-
lin delivery have been published with moderately encouraging
results [48–51]; however, there have been no large outpatient
Patient-Reported Outcomes trials to date and mean glucose appears to be lower when
using a HCL system with pre-meal announcement at this time.
In addition to improved glycemic outcomes, AP systems have While the majority of children with T1D are not using AP
been shown to decrease the burden of diabetes management systems at this time, automated insulin delivery is quickly
and increase quality of life, although this is an area in need of becoming the standard of care for those using insulin pumps,
further study. Barnard et al. reported reduced worry, improved and it is possible that AP systems will become the dominant
overnight control leading to improved daily functioning, and approach to diabetes control for all people with T1D over
improved sleep in AP users, although technical and connec- time.
tivity issues and intrusive alarms were cited as negative expe-
riences [45]. In the field of pediatrics, these outcomes are of
interest not only for children with T1D but also for their par- Conclusion
ents. Studies of parents of young children with T1D on AP
systems have reported reduced burden of diabetes manage- Diabetes technology has advanced significantly over the past
ment, less worry, and improved quality of sleep [46]. The decade, allowing for improved therapies for people living with
durability of these effects in longer-term studies remains to type 1 diabetes and hope for better quality of life, less burden
be seen. of care, and decreased risk of diabetes-related complications.
Hybrid closed-loop AP systems have consistently been found
to be safe and effective in improving glycemic outcomes,
Barriers to AP Use particularly overnight, across all populations studied including
young children. As algorithms become more advanced, the
Despite documented improved glycemic outcomes with use of ultimate goal of a fully automated system feels within reach.
diabetes technology, barriers to use remain. A recent study by
Messer et al. noted that cost and insurance coverage were the Compliance with Ethical Standards
most commonly cited barriers to the use of technology in over
400 adolescents surveyed with T1D [47]. Additional signifi- Conflict of Interest MJS has had grant support from Tandem Diabetes
cant barriers reported included the hassle of wearing devices Care, Inc.; Medtronic, PLC; and Insulet Corporation. MDD has had grant
and disliking the device on their body. Additionally, some or material support from Tandem Diabetes Care, Inc.; Medtronic, PLC;
and DexCom, Inc.
adolescent subjects reported that they were nervous that the
device might not work adequately or safely, that it would Human and Animal Rights and Informed Consent This article does not
actually increase the time required to manage their diabetes, contain any studies with human or animal subjects performed by any of
and that using the system would influence how others individ- the authors.
uals think of them.

References
Future Directions
Papers of particular interest, published recently, have been
Postprandial hyperglycemia is responsible for the majority of
highlighted as:
time spent out of range on HCL systems. In addition to devel-
• Of importance
oping accurate meal-detection algorithms to achieve a fully
•• Of major importance
automated system, improved insulin analogs with a more rap-
id onset and a shorter duration of action will likely be neces- 1. Mobasseri M, Shirmohammadi M, Amiri T, Vahed N, Hosseini
sary to improve control of postprandial glycemic excursions. Fard H, Ghojazadeh M. Prevalence and incidence of type 1 diabetes
As previously noted, the amylin analogue pramlintide is also in the world: a systematic review and meta-analysis. Health Promot
being studied as a method to decrease glycemic excursions Perspect. 2020;10:98–115.
2. Nathan DM, Genuth S, Lachin J, Cleary P, Crofford O, Davis M,
related to meals. Additionally, work is being done to allow et al. The effect of intensive treatment of diabetes on the develop-
for increased individualization and adaptation of AP systems ment and progression of long-term complications in insulin-
to changes in insulin sensitivity throughout the day. dependent diabetes mellitus. N Engl J Med. 1993;329:977–86.
Curr Nutr Rep (2021) 10:47–57 55

3. Children and adolescents: standards of medical care in diabetes- controlled trial in adolescents with type 1 diabetes. Diabetes Care.
2020. Diabetes Care 2020:43:S163-s182 2018;41:303–10.
4. Di Meglio LA, Acerini CL, Codner E, Craig ME, Hofer SE, Pillay 19. Bergenstal RM, Garg S, Weinzimer SA, Buckingham BA, Bode
K, et al. ISPAD Clinical Practice Consensus Guidelines 2018: BW, Tamborlane WV, et al. Safety of a hybrid closed-loop insulin
Glycemic control targets and glucose monitoring for children, ado- delivery system in patients with type 1 diabetes. Jama. 2016;316:
lescents, and young adults with diabetes. Pediatr Diabetes. 2018(19 1407–8.
Suppl 27):105–14. 20. Garg SK, Weinzimer SA, Tamborlane WV, Buckingham BA,
5.• Foster NC, Beck RW, Miller KM, Clements MA, Rickels MR, LA Bode BW, Bailey TS, et al. Glucose outcomes with the in-home
DM, et al. State of type 1 diabetes management and outcomes from use of a hybrid closed-loop insulin delivery system in adolescents
the T1D Exchange in 2016-2018. Diabetes Technol Ther. and adults with type 1 diabetes. Diabetes Technol Ther. 2017;19:
2019;21:66–72. This report from a large diabetes registry 155–63.
(T1D Exchange) noted that while use of diabetes technology 21.•• Forlenza GP, Pinhas-Hamiel O, Liljenquist DR, Shulman DI,
has increased in children with T1D, mean HbA1c has Bailey TS, Bode BW, et al. Safety evaluation of the MiniMed
remained relatively stable over the past 6 years and appears 670G system in children 7-13 years of age with type 1 diabetes.
to have actually worsened in adolescents. Diabetes Technol Ther. 2019;21:11–9. This pivotal trial on the
6. Doyle FJ 3rd, Huyett LM, Lee JB, Zisser HC, Dassau E. Closed- use of the Medtronic 670G system in school-aged children with
loop artificial pancreas systems: engineering the algorithms. T1D showed increased TIR and lower HbA1c over 3 months of
Diabetes Care. 2014;37:1191–7. use and led to FDA approval of the device in this age range.
7. Heinemann L, Schoemaker M, Schmelzeisen-Redecker G, 22. Stone MP, Agrawal P, Chen X, Liu M, Shin J, Cordero TL, et al.
Hinzmann R, Kassab A, Freckmann G, et al. Benefits and limita- Retrospective analysis of 3-month real-world glucose data after the
tions of MARD as a performance parameter for continuous glucose MiniMed 670G system commercial launch. Diabetes Technol Ther.
monitoring in the interstitial space. J Diabetes Sci Technol. 2018;20:689–92.
2020;14:135–50.
23. Berget C, Messer LH, Vigers T, Frohnert BI, Pyle L, Wadwa RP,
8. Kovatchev BP, Patek SD, Ortiz EA, Breton MD. Assessing sensor
et al. Six months of hybrid closed loop in the real-world: an evalu-
accuracy for non-adjunct use of continuous glucose monitoring.
ation of children and young adults using the 670G system. Pediatr
Diabetes Technol Ther. 2015;17:177–86.
Diabetes. 2020;21:310–8.
9. Tauschmann M, Hermann JM, Freiberg C, Papsch M, Thon A,
Heidtmann B, et al. Reduction in diabetic ketoacidosis and severe 24. Salehi P, Roberts AJ, Kim GJ. Efficacy and safety of real-life usage
hypoglycemia in pediatric type 1 diabetes during the first year of of MiniMed 670G automode in children with type 1 diabetes less
continuous glucose monitoring: a multicenter analysis of 3,553 than 7 years old. Diabetes Technol Ther. 2019;21:448–51.
subjects from the DPV registry. Diabetes Care. 2020;43. United 25.•• Brown SA, Kovatchev BP, Raghinaru D, Lum JW, Buckingham
States:e40–2. BA, Kudva YC, et al. Six-month randomized, multicenter trial of
10. Battelino T, Danne T, Bergenstal RM, Amiel SA, Beck R, Biester closed-loop control in type 1 diabetes. N Engl J Med. 2019;381:
T, et al. Clinical targets for continuous glucose monitoring data 1707–17. This pivotal trial on the use of the Tandem Control-IQ
interpretation: recommendations from the international consensus system in adolescents and adults with T1D demonstrated in-
on time in range. Diabetes Care. 2019;42:1593–603. creased TIR and lower HbA1c on the hybrid closed-loop sys-
11. Lal RA, Ekhlaspour L, Hood K, Buckingham B. Realizing a tem compared to standard therapy and led to FDA approval of
closed-loop (artificial pancreas) system for the treatment of type 1 the device.
diabetes. Endocr Rev. 2019;40:1521–46. 26.•• Breton MD, Kanapka LG, Beck RW, Ekhlaspour L, Forlenza GP,
12. Van Name MA, Hilliard ME, Boyle CT, Miller KM, DeSalvo DJ, Cengiz E, et al. A randomized trial of closed-loop control in chil-
Anderson BJ, et al. Nighttime is the worst time: parental fear of dren with type 1 diabetes. N Engl J Med. 2020;383:836–45. This
hypoglycemia in young children with type 1 diabetes. Pediatr pivotal trial on the use of the Tandem Control-IQ system in
Diabetes. 2018;19:114–20. school-aged children with T1D demonstrated increased TIR
13. Agrawal P, Zhong A, Welsh JB, Shah R, Kaufman FR. on the hybrid closed-loop system compared to standard thera-
Retrospective analysis of the real-world use of the threshold sus- py, resulting in FDA approval of the device in this age range.
pend feature of sensor-augmented insulin pumps. Diabetes 27. Pinsker JE, Müller L, Constantin A, Leas S, Manning M, McElwee
Technol Ther. 2015;17:316–9. Malloy M, Singh H, Habif S. Real-world patient reported outcomes
14. Bergenstal RM, Klonoff DC, Garg SK, Bode BW, Meredith M, and glycemic results with initiation of control-iq technology.
Slover RH, et al. Threshold-based insulin-pump interruption for Diabetes Technol Ther 2020. https://doi.org/10.1089/dia.2020.
reduction of hypoglycemia. N Engl J Med. 2013;369:224–32. 0388.
15. Zhong A, Choudhary P, McMahon C, Agrawal P, Welsh JB, 28. Lee MH, Vogrin S, Paldus B, Jones HM, Obeyesekere V, Sims C,
Cordero TL, et al. Effectiveness of automated insulin management et al. Glucose control in adults with type 1 diabetes using a
features of the MiniMed(®) 640G sensor-augmented insulin pump. medtronic prototype enhanced-hybrid closed-loop system: a feasi-
Diabetes Technol Ther. 2016;18:657–63. bility study. Diabetes Technol Ther. 2019;21:499–506.
16. Battelino T, Nimri R, Dovc K, Phillip M, Bratina N. Prevention of 29. de Bock M, Dart J, Hancock M, Smith G, Davis EA, Jones TW.
hypoglycemia with predictive low glucose insulin suspension in Performance of medtronic hybrid closed-loop iterations: results
children with type 1 diabetes: a randomized controlled trial. from a randomized trial in adolescents with type 1 diabetes.
Diabetes Care. 2017;40:764–70. Diabetes Technol Ther. 2018;20:693–7.
17. Forlenza GP, Li Z, Buckingham BA, Pinsker JE, Cengiz E, Wadwa 30. Paldus B, Lee MH, Jones HM, McAuley SA, Horsburgh JC, Roem
RP, et al. Predictive low-glucose suspend reduces hypoglycemia in KL, et al. Glucose control using a standard versus an enhanced
adults, adolescents, and children with type 1 diabetes in an at-home hybrid closed loop system: a randomized crossover study.
randomized crossover study: results of the PROLOG trial. Diabetes Diabetes Technol Ther. 2019;21:56–8.
Care. 2018;41:2155–61. 31. Jennings P, Hussain S. Do-it-yourself artificial pancreas systems: a
18. Abraham MB, Nicholas JA, Smith GJ, Fairchild JM, King BR, review of the emerging evidence and insights for healthcare profes-
Ambler GR, et al. Reduction in hypoglycemia with the predictive sionals. J Diabetes Sci Technol. 2020;14:868–77.
low-glucose management system: a long-term randomized
56 Curr Nutr Rep (2021) 10:47–57

32. Duke MD, Fredlock AA. Do-it-yourself (DIY) systems in diabetes: 47. Messer LH, Tanenbaum ML, Cook PF, Wong JJ, Hanes SJ,
a family and provider perspective. J Diabetes Sci Technol. 2020;14: Driscoll KA, et al. Cost, hassle, and on-body experience: barriers
917–21. to diabetes device use in adolescents and potential intervention
33. Braune K, O’Donnell S, Cleal B, Lewis D, Tappe A, Willaing I, targets. Diabetes Technol Ther. 2020;22:760–7.
et al. Real-world use of do-it-yourself artificial pancreas systems in 48. Cameron FM, Ly TT, Buckingham BA, Maahs DM, Forlenza GP,
children and adolescents with type 1 diabetes: online survey and Levy CJ, et al. Closed-loop control without meal announcement in
analysis of self-reported clinical outcomes. JMIR Mhealth Uhealth. type 1 diabetes. Diabetes Technol Ther. 2017;19:527–32.
2019;7:e14087. 49. Forlenza GP, Cameron FM, Ly TT, Lam D, Howsmon DP, Baysal
34. Palmer W, Greeley SAW, Letourneau-Freiberg LR, Naylor RN. N, et al. Fully closed-loop multiple model probabilistic predictive
Using a do-it-yourself artificial pancreas: perspectives from patients controller artificial pancreas performance in adolescents and adults
and diabetes providers. J Diabetes Sci Technol. 2020;14:860–7. in a supervised hotel setting. Diabetes Technol Ther. 2018;20:335–
35. Russell SJ, El-Khatib FH, Sinha M, Magyar KL, McKeon K, 43.
Goergen LG, et al. Outpatient glycemic control with a bionic pan- 50. Weinzimer SA, Steil GM, Swan KL, Dziura J, Kurtz N,
creas in type 1 diabetes. N Engl J Med. 2014;371:313–25. Tamborlane WV. Fully automated closed-loop insulin delivery
36. Russell SJ, Hillard MA, Balliro C, Magyar KL, Selagamsetty R, versus semiautomated hybrid control in pediatric patients with type
Sinha M, et al. Day and night glycaemic control with a bionic 1 diabetes using an artificial pancreas. Diabetes Care. 2008;31:
pancreas versus conventional insulin pump therapy in preadoles- 934–9.
cent children with type 1 diabetes: a randomised crossover trial. 51. Dovc K, Piona C, Yeşiltepe Mutlu G, Bratina N, Jenko Bizjan B,
Lancet Diabetes Endocrinol. 2016;4:233–43. Lepej D, et al. Faster compared with standard insulin aspart during
37. Sherwood JS, Jafri RZ, Balliro CA, Zheng H, El-Khatib FH, day-and-night fully closed-loop insulin therapy in type 1 diabetes: a
Damiano ER, et al. Automated glycemic control with the bionic double-blind randomized crossover trial. Diabetes Care. 2020;43:
pancreas in cystic fibrosis-related diabetes: a pilot study. J Cyst 29–36.
Fibros. 2020;19:159–61. 52. Ekhlaspour L, Forlenza GP, Chernavvsky D, Maahs DM, Wadwa
RP, Deboer MD, Messer LH, Town M, Pinnata J, Kruse G,
38.• El-Khatib FH, Balliro C, Hillard MA, Magyar KL, Ekhlaspour L,
Kovatchev BP, Buckingham BA, Breton MD. Closed loop control
Sinha M, et al. Home use of a bihormonal bionic pancreas versus
in adolescents and children during winter sports: Use of the Tandem
insulin pump therapy in adults with type 1 diabetes: a multicentre
Control-IQ AP system. Pediatr Diabetes. 2019:20:759–68.
randomised crossover trial. Lancet (London, England). 2017;389:
53. Forlenza GP, Ekhlaspour L, Breton M, Maahs DM, Wadwa RP,
369–80. This study reported lower mean glucose in adult sub-
DeBoer M, et al. Successful at-home use of the Tandem Control-IQ
jects using a bihormonal (insulin and glucagon) fully automated
bionic pancreas without need for carbohydrate counting com- artificial pancreas system in young children during a randomized
pared to standard therapy. However, more nausea was report- controlled trial. Diabetes Technol Ther. 2019;21:159–69.
ed in the experimental group. 54. Schoelwer MJ, Robic JL, Gautier T, Fabris C, Carr K, Clancy-
Oliveri M, et al. Safety and efficacy of initializing the Control-IQ
39. Haidar A, Tsoukas MA, Bernier-Twardy S, Yale JF, Rutkowski J,
artificial pancreas system based on total daily insulin in adolescents
Bossy A, et al. A novel dual-hormone insulin-and-pramlintide arti-
with type 1 diabetes. Diabetes Technol Ther. 2020;22:594–601.
ficial pancreas for type 1 diabetes: a randomized controlled cross-
55. Kovatchev BP, Kollar L, Anderson SM, Barnett C, Breton MD,
over trial. Diabetes Care. 2020;43:597–606.
Carr K, et al. Evening and overnight closed-loop control versus
40. Keith-Hynes P, Guerlain S, Mize B, Hughes-Karvetski C, Khan M,
24/7 continuous closed-loop control for type 1 diabetes: a
McElwee-Malloy M, et al. DiAs user interface: a patient-centric
randomised crossover trial. Lancet Digit Health. 2020;2:e64–73.
interface for mobile artificial pancreas systems. J Diabetes Sci
56. Lal RA, Basina M, Maahs DM, Hood K, Buckingham B, Wilson
Technol. 2013;7:1416–26.
DM. One year clinical experience of the first commercial hybrid
41. Anderson SM, Raghinaru D, Pinsker JE, Boscari F, Renard E, closed-loop system. Diabetes Care. 2019;42:2190–6.
Buckingham BA, et al. Multinational home use of closed-loop con- 57. Messer LH, Berget C, Vigers T, Pyle L, Geno C, Wadwa RP, et al.
trol is safe and effective. Diabetes Care. 2016;39:1143–50. Real world hybrid closed-loop discontinuation: predictors and per-
42. Tauschmann M, Thabit H, Bally L, Allen JM, Hartnell S, Wilinska ceptions of youth discontinuing the 670G system in the first 6
ME, et al. Closed-loop insulin delivery in suboptimally controlled months. Pediatr Diabetes. 2020;21:319–27.
type 1 diabetes: a multicentre, 12-week randomised trial. Lancet 58. Tauschmann M, Allen JM, Nagl K, Fritsch M, Yong J, Metcalfe E,
(London, England). 2018;392:1321–9. et al. Home use of day-and-night hybrid closed-loop insulin deliv-
43. Deshpande S, Pinsker JE, Zavitsanou S, Shi D, Tompot R, ery in very young children: a multicenter, 3-week, randomized trial.
Church MM, et al. Design and clinical evaluation of the Diabetes Care. 2019;42:594–600.
Interoperable Artificial Pancreas System (iAPS) smartphone 59. Buckingham BA, Christiansen MP, Forlenza GP, Wadwa RP,
app: interoperable components with modular design for pro- Peyser TA, Lee JB, et al. Performance of the Omnipod personalized
gressive artificial pancreas research and development. model predictive control algorithm with meal bolus challenges in
Diabetes Technol Ther. 2019;21:35–43. adults with type 1 diabetes. Diabetes Technol Ther. 2018;20:585–
44. Kovatchev B, Anderson SM, Raghinaru D, Kudva YC, Laffel LM, 95.
Levy C, et al. Randomized controlled trial of mobile closed-loop 60. Buckingham BA, Forlenza GP, Pinsker JE, Christiansen MP,
control. Diabetes Care. 2020;43:607–15. Wadwa RP, Schneider J, et al. Safety and feasibility of the
45. Barnard KD, Venkat MV, Close K, Heinemann L, Weissberg- OmniPod hybrid closed-loop system in adult, adolescent, and pe-
Benchell J, Hood KK, et al. PsychDT working group: report psy- diatric patients with type 1 diabetes using a personalized model
chosocial aspects of artificial pancreas systems. J Diabetes Sci predictive control algorithm. Diabetes Technol Ther. 2018;20:
Technol. 2015;9:925–8. 257–62.
46. Musolino G, Dovc K, Boughton CK, Tauschmann M, Allen JM, 61. Forlenza GP, Buckingham BA, Christiansen MP, Wadwa RP,
Nagl K, et al. Reduced burden of diabetes and improved quality of Peyser TA, Lee JB, et al. Performance of Omnipod personalized
life: experiences from unrestricted day-and-night hybrid closed- model predictive control algorithm with moderate intensity exercise
loop use in very young children with type 1 diabetes. Pediatr in adults with type 1 diabetes. Diabetes Technol Ther. 2019;21:
Diabetes. 2019;20:794–9. 265–72.
Curr Nutr Rep (2021) 10:47–57 57

62. Sherr JL, Buckingham BA, Forlenza GP, Galderisi A, Ekhlaspour results of early phase feasibility studies. Diabetes Technol Ther.
L, Wadwa RP, et al. Safety and performance of the Omnipod hybrid 2020.
closed-loop system in adults, adolescents, and children with type 1 65. Castle JR, El Youssef J, Wilson LM, Reddy R, Resalat N, Branigan
diabetes over 5 days under free-living conditions. Diabetes Technol D, et al. Randomized outpatient trial of single- and dual-hormone
Ther. 2020;22:174–84. closed-loop systems that adapt to exercise using wearable sensors.
63. Ekhlaspour L, Nally LM, El-Khatib FH, Ly TT, Clinton P, Frank E, Diabetes Care. 2018;41:1471–7.
et al. Feasibility studies of an insulin-only bionic pancreas in a
home-use setting. J Diabetes Sci Technol. 2019;13:1001–7.
Publisher’s Note Springer Nature remains neutral with regard to jurisdic-
64. Christiansen M, Bartee A, Lalonde A, Jones RE, Katz M, Wolpert
tional claims in published maps and institutional affiliations.
H, et al. Performance of an automated insulin delivery system:

You might also like